Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 6 de 6
Filter
Add more filters










Database
Language
Publication year range
1.
Clin Cancer Res ; 10(23): 7842-51, 2004 Dec 01.
Article in English | MEDLINE | ID: mdl-15585616

ABSTRACT

The anti-CD20 antibody rituximab is useful in the treatment of certain B-cell malignancies, most notably non-Hodgkin's lymphoma. Its efficacy has been increased when used in combination with chemotherapy, yet anti-CD20 monoclonal antibodies (mAbs) directly conjugated with drugs such as doxorubicin (Dox) have failed to deliver drug or to demonstrate antitumor activity. We have produced anti-CD20 antibody-drug conjugates that possess potent antitumor activity by using the anti-mitotic agent, monomethyl auristatin E (MMAE), linked via the lysosomally cleavable dipeptide, valine-citrulline (vc). Two anti-CD20 conjugates, rituximab-vcMMAE and 1F5-vcMMAE, were selectively cytotoxic against CD20(+) B-lymphoma cell lines, with IC(50) values ranging from 50 ng/mL to 1 microg/mL. Unlike rituximab, which showed diffuse surface localization, rituximab-vcMMAE capped and was internalized within 4 hours after binding to CD20(+) B cells. Internalization of rituximab-vcMMAE was followed by rapid G(2)-M phase arrest and onset of apoptosis. Anti-CD20 antibody-drug conjugates prepared with Dox were internalized and localized as with rituximab-vcMMAE, yet these were not effective for drug delivery (IC(50) > 50 microg/mL). Consistent with in vitro activity, rituximab-vcMMAE showed antitumor efficacy in xenograft models of CD20-positive lymphoma at doses where rituximab or rituximab-Dox conjugates were ineffective. These data indicate that anti-CD20-based antibody-drug conjugates are effective antitumor agents when prepared with a stable, enzyme-cleavable peptide linkage to highly potent cytotoxic agents such as MMAE.


Subject(s)
Antibodies, Monoclonal/pharmacology , Antigens, CD20/immunology , Antineoplastic Agents/pharmacology , Lymphoma, B-Cell , Oligopeptides/metabolism , Oligopeptides/pharmacology , Animals , Antibodies, Monoclonal/metabolism , Antibodies, Monoclonal, Murine-Derived , Antineoplastic Agents/metabolism , Apoptosis/drug effects , Cell Division/drug effects , Doxorubicin/pharmacology , Drug Delivery Systems , G2 Phase/drug effects , Humans , Lymphoma, B-Cell/drug therapy , Lymphoma, B-Cell/immunology , Mice , Oligopeptides/adverse effects , Oligopeptides/chemistry , Rituximab , Tumor Cells, Cultured , Xenograft Model Antitumor Assays
2.
Clin Cancer Res ; 10(20): 7063-70, 2004 Oct 15.
Article in English | MEDLINE | ID: mdl-15501986

ABSTRACT

PURPOSE: An antibody-drug conjugate consisting of monomethyl auristatin E (MMAE) conjugated to the anti-CD30 monoclonal antibody (mAb) cAC10, with eight drug moieties per mAb, was previously shown to have potent cytotoxic activity against CD30(+) malignant cells. To determine the effect of drug loading on antibody-drug conjugate therapeutic potential, we assessed cAC10 antibody-drug conjugates containing different drug-mAb ratios in vitro and in vivo. EXPERIMENTAL DESIGN: Coupling MMAE to the cysteines that comprise the interchain disulfides of cAC10 created an antibody-drug conjugate population, which was purified using hydrophobic interaction chromatography to yield antibody-drug conjugates with two, four, and eight drugs per antibody (E2, E4, and E8, respectively). Antibody-drug conjugate potency was tested in vitro against CD30(+) lines followed by in vivo xenograft models. The maximum-tolerated dose and pharmacokinetic profiles of the antibody-drug conjugates were investigated in mice. RESULTS: Although antibody-drug conjugate potency in vitro was directly dependent on drug loading (IC(50) values E8

Subject(s)
Antibodies, Monoclonal/immunology , Immunoconjugates/immunology , Lymphoma, Large B-Cell, Diffuse/immunology , Oligopeptides/pharmacology , Animals , Antibodies, Monoclonal/pharmacokinetics , Antibodies, Monoclonal/pharmacology , Humans , Immunoconjugates/pharmacokinetics , Immunoconjugates/pharmacology , Ki-1 Antigen/immunology , Lymphoma, Large B-Cell, Diffuse/pathology , Maximum Tolerated Dose , Mice , Oligopeptides/pharmacokinetics , Transplantation, Heterologous , Tumor Cells, Cultured
3.
Bioconjug Chem ; 14(5): 860-9, 2003.
Article in English | MEDLINE | ID: mdl-13129388

ABSTRACT

The L49 single-chain Fv fused to beta-lactamase (L49-sFv-bL) combined with the prodrug C-Mel is an effective anticancer agent against tumor cells expressing the p97 antigen. However, large-scale production of L49-sFv-bL from refolded E. coli inclusion bodies has been problematic due to inefficient refolding and instability of the fusion protein. Sequence analysis of the L49-sFv framework regions revealed three residues in the framework regions at positions L2, H82B, and H91, which are not conserved for their position, occurring in <1% of sequences in Fv sequence databases. One further unusual residue, found in <3% of variable sequences, was observed at position H39. Each unusual residue was mutated to a conserved residue for its position and tested for refolding yield from inclusion bodies following expression in E. coli. The three V(H) single mutants showed improvement in the yield of active protein and were combined to form double and triple mutants resulting in a 7-8-fold increased yield compared to the parental protein. In an attempt to further improve yield, the orientation of the triple mutant was reversed to create a bL-L49-sFv fusion protein resulting in a 3-fold increase in expressed inclusion body protein and producing a 20-fold increase in the yield of purified protein compared to the parental protein. The triple mutants in both orientations displayed increased stability in murine plasma and binding affinity was not affected by the introduced mutations. Both triple mutants also displayed potent in vitro cytotoxicity and in vivo antitumor activity against p97 expressing melanoma cells and tumor xenografts, respectively. These results show that a rational protein-engineering approach improved the yield, stability, and refolding characteristics of L49-sFv-bL while maintaining binding affinity and therapeutic efficacy.


Subject(s)
Antibodies, Neoplasm/biosynthesis , Antibodies, Neoplasm/genetics , Antineoplastic Agents/metabolism , Immunoglobulin G/biosynthesis , Immunoglobulin G/genetics , Prodrugs/metabolism , Protein Engineering/methods , Recombinant Fusion Proteins/biosynthesis , Recombinant Fusion Proteins/genetics , beta-Lactamases/biosynthesis , beta-Lactamases/genetics , Animals , Antibodies, Monoclonal/genetics , Antibodies, Monoclonal/therapeutic use , Antibodies, Neoplasm/administration & dosage , Antibodies, Neoplasm/metabolism , Antigens, Neoplasm , Antineoplastic Agents/administration & dosage , Antineoplastic Agents/chemistry , Binding Sites, Antibody/genetics , Dose-Response Relationship, Drug , Drug Stability , Female , Humans , Immunoglobulin G/administration & dosage , Immunoglobulin G/metabolism , Melanoma-Specific Antigens , Mice , Mice, Inbred BALB C , Mutagenesis, Site-Directed , Neoplasm Proteins/genetics , Neoplasm Proteins/immunology , Prodrugs/administration & dosage , Prodrugs/chemistry , Recombinant Fusion Proteins/administration & dosage , Recombinant Fusion Proteins/metabolism , Tumor Cells, Cultured , Xenograft Model Antitumor Assays/methods , beta-Lactamases/administration & dosage , beta-Lactamases/metabolism
4.
Nat Biotechnol ; 21(7): 778-84, 2003 Jul.
Article in English | MEDLINE | ID: mdl-12778055

ABSTRACT

We describe the in vitro and in vivo properties of monoclonal antibody (mAb)-drug conjugates consisting of the potent synthetic dolastatin 10 analogs auristatin E (AE) and monomethylauristatin E (MMAE), linked to the chimeric mAbs cBR96 (specific to Lewis Y on carcinomas) and cAC10 (specific to CD30 on hematological malignancies). The linkers used for conjugate formation included an acid-labile hydrazone and protease-sensitive dipeptides, leading to uniformly substituted conjugates that efficiently released active drug in the lysosomes of antigen-positive (Ag+) tumor cells. The peptide-linked mAb-valine-citrulline-MMAE and mAb-phenylalanine-lysine-MMAE conjugates were much more stable in buffers and plasma than the conjugates of mAb and the hydrazone of 5-benzoylvaleric acid-AE ester (AEVB). As a result, the mAb-Val-Cit-MMAE conjugates exhibited greater in vitro specificity and lower in vivo toxicity than corresponding hydrazone conjugates. In vivo studies demonstrated that the peptide-linked conjugates induced regressions and cures of established tumor xenografts with therapeutic indices as high as 60-fold. These conjugates illustrate the importance of linker technology, drug potency and conjugation methodology in developing safe and efficacious mAb-drug conjugates for cancer therapy.


Subject(s)
Adenocarcinoma/drug therapy , Adenocarcinoma/pathology , Antibodies, Monoclonal/chemistry , Antibodies, Monoclonal/therapeutic use , Antineoplastic Agents/chemistry , Antineoplastic Agents/therapeutic use , Oligopeptides/chemistry , Oligopeptides/therapeutic use , Animals , Antibodies, Monoclonal/adverse effects , Antineoplastic Agents/adverse effects , Antineoplastic Agents/chemical synthesis , Cells, Cultured/drug effects , Drug Delivery Systems/methods , Drug Stability , Humans , Immunotoxins/adverse effects , Immunotoxins/chemistry , Immunotoxins/therapeutic use , Mice , Mice, Inbred BALB C , Mice, Nude , Mice, SCID , Neoplasm Transplantation , Oligopeptides/adverse effects , Oligopeptides/chemical synthesis , Protein Binding , Treatment Outcome
5.
Blood ; 102(4): 1458-65, 2003 Aug 15.
Article in English | MEDLINE | ID: mdl-12714494

ABSTRACT

The chimeric monoclonal antibody cAC10, directed against CD30, induces growth arrest of CD30+ cell lines in vitro and has pronounced antitumor activity in severe combined immunodeficiency (SCID) mouse xenograft models of Hodgkin disease. We have significantly enhanced these activities by conjugating to cAC10 the cytotoxic agent monomethyl auristatin E (MMAE) to create the antibody-drug conjugate cAC10-vcMMAE. MMAE, a derivative of the cytotoxic tubulin modifier auristatin E, was covalently coupled to cAC10 through a valine-citrulline peptide linker. The drug was stably attached to the antibody, showing only a 2% release of MMAE following 10-day incubation in human plasma, but it was readily cleaved by lysosomal proteases after receptor-mediated internalization. Release of MMAE into the cytosol induced G2/M-phase growth arrest and cell death through the induction of apoptosis. In vitro, cAC10-vcMMAE was highly potent and selective against CD30+ tumor lines (IC50 less than 10 ng/mL) but was more than 300-fold less active on antigen-negative cells. In SCID mouse xenograft models of anaplastic large cell lymphoma or Hodgkin disease, cAC10-vcMMAE was efficacious at doses as low as 1 mg/kg. Mice treated at 30 mg/kg cAC10-vcMMAE showed no signs of toxicity. These data indicate that cAC10-vcMMAE may be a highly effective and selective therapy for the treatment of CD30+ neoplasias.


Subject(s)
Antibodies, Monoclonal/pharmacology , Antineoplastic Agents/pharmacology , Immunoconjugates/pharmacology , Immunotoxins/pharmacology , Ki-1 Antigen/immunology , Oligopeptides/pharmacology , Animals , Antibodies, Monoclonal/adverse effects , Antibodies, Monoclonal/chemistry , Antineoplastic Agents/adverse effects , Antineoplastic Agents/chemistry , Apoptosis/drug effects , Brentuximab Vedotin , Cell Cycle/drug effects , Drug Stability , Hodgkin Disease/drug therapy , Hodgkin Disease/immunology , Hodgkin Disease/metabolism , Humans , Immunoconjugates/chemistry , Immunotoxins/chemistry , Inhibitory Concentration 50 , Ki-1 Antigen/metabolism , Mice , Mice, SCID , Oligopeptides/adverse effects , Oligopeptides/chemistry , Tumor Cells, Cultured , Xenograft Model Antitumor Assays
6.
Cancer Res ; 62(13): 3736-42, 2002 Jul 01.
Article in English | MEDLINE | ID: mdl-12097283

ABSTRACT

The leukocyte activation marker CD30 is highly expressed on the Reed Sternberg cells of Hodgkin's disease (HD). On normal tissues, CD30 has a restricted expression profile limited to activated T cells, activated B cells, and activated natural killer cells. This expression profile makes CD30 an ideal target for monoclonal antibody (mAb)-based therapies of Hodgkin's disease. CD30 mAbs have been shown to be effective in in vitro and in vivo models of hematologic malignancies such as anaplastic large cell lymphoma, yet these mAb have not been efficacious in HD models. We have found that a mAb against CD30, AC10, was able to inhibit the growth of HD cell lines in vitro. To generate a more clinically relevant molecule, the variable regions from AC10 were cloned into an expression construct containing the human gamma1 heavy chain and kappa light chain constant regions. The resulting chimeric antibody, designated SGN-30, retained the binding and in vitro growth-inhibitory activities of the parental antibody. Treatment of HD cell lines with SGN-30 in vitro resulted in growth arrest in the G(1) phase of the cell cycle and DNA fragmentation consistent with apoptosis in the HD line L540cy. Severe combined immunodeficient mouse xenograft models of disseminated HD treated with SGN-30 produced significant increases in survival. Similarly, xenograft models of localized HD demonstrated dose-dependent reduction in tumor mass in response to SGN-30 therapy. SGN-30 is being developed for the treatment of patients who have HD that is refractory to initial treatment or who have relapsed and have limited therapeutic options.


Subject(s)
Antibodies, Monoclonal/pharmacology , DNA Fragmentation , Hodgkin Disease/therapy , Ki-1 Antigen/immunology , Animals , Antibodies, Monoclonal/biosynthesis , Antibodies, Monoclonal/genetics , Antibodies, Monoclonal/immunology , CHO Cells , Cell Cycle/drug effects , Cell Division/drug effects , Cricetinae , Hodgkin Disease/genetics , Hodgkin Disease/immunology , Hodgkin Disease/pathology , Humans , Hybridomas , Mice , Mice, SCID , Tumor Cells, Cultured , Xenograft Model Antitumor Assays
SELECTION OF CITATIONS
SEARCH DETAIL
...